You are on page 1of 30

Fluorescent aminoglycoside antibiotics and methods for

accurately monitoring uptake by bacteria


Mahnaz Sabeti Azad, Maho Okuda, Mélina Cyrenne, Mickael Bourge,
Marie-Pierre Heck, Satoko Yoshizawa, Dominique Fourmy

To cite this version:


Mahnaz Sabeti Azad, Maho Okuda, Mélina Cyrenne, Mickael Bourge, Marie-Pierre Heck, et al..
Fluorescent aminoglycoside antibiotics and methods for accurately monitoring uptake by bacteria.
ACS Infectious Diseases, American Chemical Society, 2020, 6 (5), pp.1008-1017. �10.1021/acsin-
fecdis.9b00421�. �hal-02516160�

HAL Id: hal-02516160


https://hal.archives-ouvertes.fr/hal-02516160
Submitted on 6 Jan 2021

HAL is a multi-disciplinary open access L’archive ouverte pluridisciplinaire HAL, est


archive for the deposit and dissemination of sci- destinée au dépôt et à la diffusion de documents
entific research documents, whether they are pub- scientifiques de niveau recherche, publiés ou non,
lished or not. The documents may come from émanant des établissements d’enseignement et de
teaching and research institutions in France or recherche français ou étrangers, des laboratoires
abroad, or from public or private research centers. publics ou privés.
Fluorescent aminoglycoside antibiotics and methods for accurately
monitoring uptake by bacteria
Mahnaz Sabeti Azad†, Maho Okuda†,‡, Mélina Cyrenne†, Mickael Bourge†, Marie-Pierre Heck§, Satoko
Yoshizawa†, and Dominique Fourmy*,†.

Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198, Gif-sur-Yvette, France.
§
Université Paris-Saclay, CEA, Service de Chimie Bio-organique et de Marquage, 91191 Gif-sur-Yvette, France.
Email: dominique.fourmy@i2bc.paris-saclay.fr
Characterizing how multidrug-resistant bacteria circumvent the action of clinically used or novel antibiotics requires a detailed
understanding of how the antibiotics interact with and cross bacterial membranes to accumulate in the cells and exert their action.
When monitoring the interactions of drugs with bacteria it remains challenging to differentiate functionally relevant internalized
drug levels from non-specific binding. Fluorescence is a method of choice for observing dynamics of biomolecules. In order to
facilitate studies involving aminoglycoside antibiotics, we have generated fluorescently labeled aminoglycoside derivatives with
uptake and bactericidal activities similar, albeit with a moderate loss, to those of the parent drug. The method combines fluores-
cence microscopy with fluorescence-activated cell sorting (FACS) using neomycin coupled to non-permeable cyanine dyes. Fluo-
rescence imaging allowed membrane-bound antibiotic to be distinguished from molecules in the cytoplasm. Patterns of uptake were
assigned to different populations in the FACS analysis. Our study illustrates how fluorescent derivatives of an aminoglycoside
enable a robust characterization of the three components of uptake: membrane binding, EDPI, and EDPII. Because EDPI levels are
weak compared to the two other types of accumulation and critical for the action of these drugs, the three components of uptake
must be taken into account separately when drawing conclusions about aminoglycoside function.

Keywords: Fluorescence; antibiotics; aminoglycosides; uptake; flow cytometry; microscopy.

Aminoglycosides are among the oldest antibiotics in clinical portant for understanding the basis of aminoglycoside action.
use. These broad spectrum antibiotics are used against clini- This has been often achieved by study of cultured cells treat-
cally important bacteria and for treatment of infections ed with radioactively labeled aminoglycosides.25,26,32 A
caused by multidrug-resistant bacteria.1,2 In the past decade, method has been reported for measuring intracellular levels
novel aminoglycosides have been developed with decreased of kanamycin that relies on an ELISA, but it requires cell
toxicity and susceptibility to resistance.2–7 Many widely used lysis.33 Novel tools making use of fluorescent antibiotics are
aminoglycosides contain a deoxystreptamine ring (2-DOS) emerging that enable characterization of the interactions of
that is essential for antimicrobial activity. The 2-DOS ami- antibiotics with bacteria at the level of organelles or even
noglycosides bind both subunits of the ribosome and inhibit whole organisms.34 For example, gentamicin and tobramycin
protein synthesis.8–10 These drugs cause miscoding 11 via a coupled to Texas Red have been widely used in this type of
direct interaction with ribosomal RNA (rRNA) of the small analysis; however this dye, by itself, penetrates cells, com-
subunit.10,12–16 Some 2-DOS aminoglycosides also interact at plicating analysis.35 Recently developed techniques make use
a specific site within the large ribosomal subunit resulting in of spectrofluorimetry, microspectrofluorimetry and kinetics
inhibition of translocation 17,18 or recycling.19 In addition, microspectrofluorimetry for single-cell analysis of naturally
aminoglycosides have been reported to inhibit initiation 20,21 fluorescent antibiotics or fluorescently-labeled com-
and perturb ribosome biogenesis.22–24 pounds.36,37 In addition, mass spectrometry was combined
Aminoglycoside uptake occurs through two energy- with spectrofluorimetry to provide insights into the efflux
dependent phases known as EDPI and EDPII.25–27 EDPI is mechanisms of fluoroquinolones.38
known to be dependent on the proton motive force (PMF).28– Depending on the analysis, the membrane-bound fraction of
30
EDPII requires active protein synthesis by aminoglyco- a drug may be quantified, biasing the measurement of true
side-sensitive ribosomes and, therefore, is energy dependent. internalized drug levels. For aminoglycosides, it is also im-
The lethal mode of action of aminoglycosides is thought to portant to differentiate EDPI and EDPII phases, as only
occur through induction of ribosomal miscoding, which EDPI leads to cell death. EDPII is a consequence of internal-
produces misfolded proteins that when inserted into the inner ization of the drug through the membrane damaged due to
membrane trigger an increase of permeability and conse- EDPI. In addition, because each cell may respond differently
quently a diffusive entry of the drug in EDPII.31 Increase in to antibiotics, an accurate description of drug uptake requires
cell killing efficiency is interpreted as an increase in amino- single-cell analysis. To finely evaluate aminoglycoside up-
glycoside uptake. Measuring aminoglycoside uptake is im- take, we developed a series of cyanine-modified neomycin
Neomycin Neo-Cy5 hydrazone (CCCP) on Neo-Cy5-induced bacterial cell death.
P. aeruginosa PA14 3.25 150 Addition of CCCP protected cells from the drug-induced
killing (Figure 3 and Figure S8), demonstrating that, as for
Measured in minimal medium MOPS-G. other aminoglycosides, the accumulation of Neo-Cy5 is PMF
Neo-Cy5 causes misreading during ribosomal decoding. dependent.
Aminoglycosides cause miscoding in vitro at concentrations 4 **** ****
in the range of 10-100 µM.5,11,43,44 Since Neo-Cy5 binds the Untreated

log 10 (survival percentage)


ribosomal decoding site on the small subunit (Figure 2(a)), 2
CCCP 30 µM
we tested the capacity of Neo-Cy5 to induce misreading
using a dual-luciferase gain-of-function assay. In this assay, 0

misreading restores the enzymatic activity of a mutant firefly


luciferase. In the mutant, codon 245, which in the wild-type -2

mRNA is CAC (coding for histidine), is replaced by the


near-cognate CGC codon (arginine).5 Translation activity -4

was analyzed by comparison to the signal due to wild-type


-6
Renilla luciferase, which served as internal standard. Like
neomycin, Neo-Cy5 caused miscoding however effects were

tic

µM
µM
io

48
1
tib
attenuated by more than one order of magnitude (Figure

in

y5
an

yc

-C
m
o
2(b)).

eo
N

eo

N
N
a) Figure 3. Neo-Cy5-induced cell killing is PMF dependent.
Neomycin
Neomycin Neomycin
Neo-Cy5 Cy5
Survival of E. coli MG1655 cells after a 4.5-h treatment with 1.5
µM neomycin or 48 µM Neo-Cy5 with and without a 2-h pre-
0.05 µM

0.05 µM

incubation with 30 µM CCCP. Plotted are values relative to


0.2 µM

0.2 µM
0.5 µM
control

0.1 µM

0.1 µM
1 µM

1 µM
5 µM

untreated control culture. Error bars are s. e. m. and were calcu-


A C G U
0

lated from three independent experiments. Significant differ-


ences, calculated by the Mann–Whitney test, are indicated (****
A1408 P < 0.0001).

G1405
A1408 b) FACS and TIRF microscopy reveal drug peripheral
Fig
(F-luc mutant) / (F-luc wt x 10-5)

accumulation and strong interaction with membranes.


21000 Neo-Cy5
Because cationic aminoglycosides can interact with the
Neomycin membranes of the bacterial cell surface, we devised a proto-
18000
col to measure only tightly-bound or internalized Neo-Cy5.
15000 We first removed weakly membrane-bound and not internal-
ized molecules by washing the cells on mixed cellulose ester
12000
filters. At least three washes with MOPS-G at 37 °C were
9000 necessary to remove non-internalized drug (Figure S9). We
measured Neo-Cy5 uptake by E. coli MG1655 cells in the
6000 exponential phase of growth in the presence of Neo-Cy5 at a
3000
concentration three times the MIC. After filtration, cells
were recovered and immediately analyzed by FACS. After a
0 short 5-minute exposure to Neo-Cy5, a signal due to the
0,01 .
0,1 1 10 100 conjugate was clearly distinguished from the background
An6bio6cs (µM) fluorescence (Figure 4(a)). We verified that the signal was
dependent on the neomycin moiety: TIRF microscopy
Figure 2. Neo-Cy5 binds ribosomal A site and induces mis- showed that the Cy5 dye alone was not incorporated into live
reading. a) Binding of neomycin (left) and Neo-Cy5 (right) to Figure 2
bacterial cells and that Cy5 did not interact strongly with
ribosomes was assayed using DMS chemical probing. Left lane membranes (Figure S10 and Figure 4(a)). Because the ob-
is a control reaction with no DMS added. Right lanes are reac-
served signal for Neo-Cy5 could correspond to molecules
tions in the presence of increasing concentrations of neomycin
tightly bound to the membrane or internalized in the
or Neo-Cy5. The protected nucleotide A1408 that belongs to the
periplasm or cytoplasm, we inspected the localization of the
neomycin binding site (Figure 1(b) is marked with an arrow. b)
Mis-incorporation of amino acids was assayed using a gain-of- drug by TIRF microscopy (Figure 4(b)). Here, periplasmic
function assay. Wild-type and mutant firefly luciferase reporters accumulation would correspond to the self-promoted mecha-
were used for in vitro translation reactions together with the nism previously proposed where aminoglycoside cross irre-
Renilla luciferase, which served as an internal control. Error versibly the outer membrane via the lipopolysaccharides in
bars are s. e. m. for three independent experiments. E. coli.45 After a 5-minute incubation, Neo-Cy5 accumulated
at the periphery of the cell and did not overlap with the fluo-
rescence of Spinach-tagged ribosomes46 (Figure 4(c) and
Neo-Cy5 has proton motive force-dependent bactericidal Figure S11)). We conclude that Neo-Cy5 bound at the mem-
activity. Aminoglycoside accumulation in bacterial cells is brane of a cell or internalized in the periplasm can readily be
known to be dependent on PMF.28 We therefore tested the
effect of the protonophore carbonyl cyanide m-chlorophenyl
derivatives that have antibacterial activity similar to the Figure 1. Structure of the Neo-Cy5 conjugate and accommoda-
parent aminoglycoside. When used with adequate protocols, tion at the ribosomal A site. a) Structure of Neo-Cy5 in which
these derivatives allow single-cell analysis of aminoglyco- Cy5 is coupled to ring III of neomycin. b) Neomycin binding
side uptake by fluorescence microscopy and fluorescence- sites in h44 of the small ribosomal subunit (blue) and in H69 of
activated cell sorting (FACS). the large ribosomal subunit (beige) (PDB code 4v52). Small and
large ribosomal proteins are shown in purple and green, respec-
tively. The red box indicates a cavity near the neomycin binding
RESULTS AND DISCUSSION site in h44 where Cy5 can be accommodated. Nucleotide A1408
Generation of Neo-cyanine derivatives. Radioactivity that is part of the neomycin binding site is indicated.
based assays25,26,32 and ELISA33 do not report on accumula- Neo-cyanine derivatives are active antibiotics. Next, we
tion at the single cell level. A fluorescent derivative with verified that Neo-Cy5 binds the decoding site of 16S rRNA
preserved uptake and bactericidal activities would facilitate in 70S ribosomes. Footprinting analysis of ribosomal parti-
analysis of uptake with single-cell resolution. We previously cles bound to Neo-Cy5 showed that the conjugate interacts
showed that the 5” position of neomycin could be modified with the decoding site of the small ribosomal subunit in a
with various functional groups without compromising anti- manner similar to neomycin and Neo-U, and like Neo-U,
microbial activity (Figure S1). Analysis of the structures of a Neo-Cy5 has slightly lower affinity than the parent antibiotic
neomycin-ribosome complex indicated that functional (Figure 2(a)). We next tested the antimicrobial activity of
groups introduced at this position are accommodated in the Neo-Cy5. The measured MIC in minimal medium was
major groove of helix 44 of the 16S rRNA in a conformation slightly higher than that of neomycin (Table 1) and similar to
that does not interfere with drug binding or induction of the MICs for Neo-U (Table S1) and closely related amino-
miscoding. glycosides.41,42 The MIC of Neo-Cy3 was also similar to that
We used a derivative of neomycin where the hydroxyl group of Neo-Cy5 (Table 1).
at the 5” position was substituted with S-(CH2)2-NH2 to MIC values were also measured in Mueller-Hinton broth for
allow introduction of extra chemical groups by coupling Gram negative and positive bacteria including ESKAPE
(Supporting information, product 5).39 Addition of a pathogens (Table 2). Tested strains included clinically rele-
thyminylacetamidoethylthio or uracil (Neo-U) at the 5” vant Gram negative pathogens such as Pseudomonas aeru-
position preserved the antimicrobial activity against E. coli ginosa, A. baumannii, K. pneumoniae, S. typhimurium, as
as measured using an MIC assay (Figure S1 and Table S1). well as the Gram positive S. aureus USA300. MIC values for
As expected, Neo-U bound to the A site on the small ribo- the parental drug neomycin were higher in rich medium
somal subunit, although affinity was slightly decreased com- when compared to minimal medium. This effect was strong-
pared to the unmodified neomycin (Figure S2). This result er for the Neo-Cy5 conjugate as MIC values were increased
indicated that large chemical modifications can be intro- by factors ranging from 20 to 40 compared to the values
duced at the 5” position of ring III without strongly perturb- measured for neomycin (Table 2). The MIC value was not
ing A-site binding or antimicrobial properties. measurable for Pseudomonas aeruginosa in rich medium and
We next labeled neomycin with sulfonated cyanines Cy3 and found to be 150 µg/ml in MOPS-G (Table 3).
Cy5 (Figures S3,S4,S5 and S6), which are non-membrane
permeable dyes suitable for fluorescence microscopy.40 The
Table 1. MIC values in µg/ml of neomycin-cyanine deriv-
spectral properties of Neo-Cy5 conjugate (Figure 1(a)) do
ative conjugates against E. coli MG1655.
not differ from those of the free dye (Figure S7). We verified
that the fluorescent probes are accommodated without any Neomycin Neo-Cy5 Neo-Cy3
steric clash by docking into the crystal structure of E. coli
M9 medium 3.0 13 ND
ribosomes bound to neomycin (PDB code 4v52). The Cy3
and Cy5 probes, which are bulkier than the uracil base, likely MOPS medium 1.1 10.4 5.2
lie in a cavity between helices h27 and h44 of 16S ribosomal Measured in minimal medium MOPS-G. ND: not determined.
RNA (Figure 1(b)).
Table 2. MIC values of Neo-Cy5 in µg/ml against Gram
es : a) b) 30S 50S
negative and positive strains.

Neomycin Neo-Cy5
E. coli MG1655 2.5 75
neomycin
P. aeruginosa PA14 2.5 > 150

Neo-Cy5Neo-Cy5
A. baumannii DSM30011 5.0 75
K. pneumoniae LM21 1.25 25
NHS Cy5 S. typhimurium14028 1.25 50
NHS Cy5 h 27
B. subtilis 0.25 6.25
* H 69 M. luteus 1.5 6.25
ation of Neo-Cy5. The primary amine group that reacts A1408
S. aureus USA300 2.5 75
ighlighted by the red circle. Other amino groups are
Neo-Cy5 h 44
foxycarbonyl
Neo-Cy5.groups.
The primary amine group that reacts Position Cy5
Measured in cation-adjusted Mueller-Hinton broth.
ted by the red circle. Other amino groups are S12
bonyl groups. Table 3. MIC values of Neo-Cy5 in µg/ml against P. aeru-
Figure 1
ginosa PA14.
detected by FACS and visualized by TIRF fluorescence gestive of EDPII accumulation. For 60 min of incubation,
microscopy. 100% of the cells had cytoplasmic uptake (n=119). Cells
exposed to Neo-Cy5 overnight showed morphological de-
fects (Figure S13) similar to what has been previously de-
Neo-Cy5 binding to bacterial membranes is saturable.
scribed for cells treated with aminoglycosides,49,50 in particu-
Binding of Neo-Cy5 to the bacterial membranes was found
lar foci formation at poles.
to be instantaneous as visualized in real time by FACS (Fig-
ure S12). Membranes of E. coli showed saturability when Addition of the protein synthesis inhibitor chloramphenicol
exposed for 5 minutes to increasing concentrations of Neo- prior to exposure to aminoglycosides decreases EDPI and
Cy5 (Figure 4(d,e)). At low concentrations, there was strong abolishes EDPII.51 We pre-incubated the cells with chloram-
heterogeneity in fluorescence levels of individual cells (Fig- phenicol before the addition of a lethal concentration of Neo-
ure 4(d)). At the highest concentrations, even though the Cy5 to evaluate the contributions of EDPI and EDPII uptake
exposure time was short to ensure that only the drug interac- by FACS (Figure 5(b)). Differences in profiles obtained with
tion with the membrane was monitored, in a fraction of cells or without chloramphenicol were very minor at 30 minutes;
high levels of uptake was observed (Figure 4(d)). Experi- TIRF imaging showed that EDPI already occurred for most
ments described in the next section demonstrated that this of the cells (Figure 5(a)). Only a slight shift of the distribu-
fraction corresponds to cells with cytoplasmic uptake. In tion toward stronger levels of fluorescence was observed
cells treated with 64 µM of Neo-Cy5, 2.9% of cells had with time. This result revealed that EDPI uptake is indeed
detectable levels of Neo-Cy5 in the cytoplasm. This value is difficult to capture by FACS. The shift of the distribution in
above the known fraction of 0.3% of cells that are dead in a absence of chloramphenicol was more pronounced at 60 and
fast growing bacterial population.47 Therefore there is rapid 150 minutes in agreement with an EDPII-driven cytoplasmic
cytoplasmic uptake at high concentrations of Neo-Cy5. uptake. We concluded that little Neo-Cy5 accumulates in the
cytoplasm via EDPI when compared to membrane-bound or
cytoplasmic levels that resulted from the EDPII-driven pro-
Neo-Cy5 is internalized mainly through EDPII. E. coli cess.
MG1655 cells were treated with 32 µM Neo-Cy5 for 30
minutes, the time period corresponding to the onset of ami-
noglycoside-induced exponential killing.48 When cells were Accumulation of Neo-Cy5 and loss of cell viability. We
imaged using TIRF, we observed that fluorescence signals next performed a detailed kinetic analysis of Neo-Cy5 up-
were distributed over the entire cell volume for most of the take at the single-cell level. Cell viability was monitored in
cells (time 30 min 88%, n=57), (Figure 5(a)). This clearly parallel. Like observed for other aminoglycosides48, Neo-
indicated cytoplasmic uptake. Most of the cells had low Cy5 displayed the characteristic lag followed by exponential
levels of fluorescence corresponding to EDPI uptake. A low killing of the cells (Figure 5(c)). The plot of the sum of the
percentage of cells had bright Cy5 fluorescence signals sug- fluorescence levels as a function of time calculated from our
100
a) Bacteria alone
b)
75
Cy5
Neo-Cy5
50
Counts

25

0
0 103 104 105 106 107
Cy5
c) BF
Cy5 +
Spinach
d) e)
0 %
cytoplasmic

0.4

1
200
Fluorescence intensity (a. u.)

spinach
160 5 0.08 %
neo-cy5

120 10 0.04 %

80 0.03 %
16
40
32 0.49 %
Counts

0
0 1 2 3 64 2.9 %
Distance (µm)
102 103 104 105
Cy5
Figure 4. Fluorescence microscopy and FACS enable detection of Neo-Cy5 bound to E. coli MG1655 cells. a) Figure 4 submitted
FACS analysis of cells
exposed to 32 µM Cy5, 32 µM Neo-Cy5, or no dye for 5 min at 37 °C. Cells were washed on a 0.45 µm-pore-size HAWP membrane filter.
Binding of Neo-Cy5 was clearly distinct from cellular autofluorescence and residual binding of the free dye. b) Representative brightfield
and TIRF microscopy images of cells incubated for 5 min with Neo-Cy5 as in panel “a”. Scale bar: 5 µm c) Neo-Cy5 does not localize
with ribosomes after a 5-minute incubation. E. coli cells expressing Spinach-tagged ribosomes were exposed to Neo-Cy5 and imaged. Left:
Brightfield. Right: Neo-Cy5 (red), Spinach (green). Bottom: plot of a cross section of fluorescence signal for a representative cell with the
peripheral pattern. d) Saturability of bacterial membranes to Neo-Cy5 binding. Cells were incubated for 5 min at 37 °C with increasing
concentrations of Neo-Cy5, filtered, and analyzed by FACS. The percentage of cells with cytoplasmic uptake was calculated from the
number of cell counts with a Cy5 signal higher than the red threshold line divided by the total number of cell counts. e) Binding curve of
the membrane-bound Neo-Cy5.
single-cell analysis recapitulated previous data obtained on the membrane-bound fraction (ratio of medians 120 min/5
cell ensembles.26 During the first 20 minutes of incubation min) (Figure 5(c)). Finally, at the beginning of the transition
with the drug, fluorescence levels increased slightly while time point (40 minutes), the cytoplasmic drug levels that
cell survival remained intact. During this period, Neo-Cy5 accumulated during EDPI, accounted for approximately one
localized at the cell periphery (Supplementary Figure 13). At third of the total levels, the other two thirds corresponded to
40 minutes, the cytoplasmic pattern became dominant with membrane/periplasmic interaction or accumulation (Figure
stronger drug accumulation (Figure 5(c,d) and Figure S14) 5(c)).
concomitantly with the onset of the decrease in cell survival
rate (Figure 5(c)). This period corresponded to a transition
CONCLUSIONS
between EDPI and EDPII. The onset of EDPII was previous-
ly considered to be the first time point at which uptake be- With the purpose of improving methodologies for measuring
comes linear and rapid.26 This time point for Neo-Cy5 was aminoglycoside uptake by bacteria we used click chemistry
around 50 minutes (Figure 5(c)). During the 20-minute tran- to develop fluorescent conjugates of neomycin that have
sition period (40 to 60-min) 90% of the cells lost viability uptake and bactericidal properties similar to the ones of the
with a sharp increase in drug uptake. After 80 minutes, 99% parent aminoglycoside. We coupled sulfonated cyanine dyes
of the cells were dead and levels of accumulation continued that are not membrane-permeable to neomycin allowing
to increase steeply and linearly during 40 minutes, reaching a clear visualization of the uptake. The obtained Neo-Cy3 and
factor of approximately 10 of increase taking as a reference Neo-Cy5 derivatives exhibited uptake properties strictly
a) a)a) 1
1
b)b)100
b) 100 2 min
2 min

50 50
2 2
1 0
1 100 0
30 min
100 30 min
2 EDPI
3 50
2 EDPI
2 min 3 3 50
0
100 60 min
2 min 3
0 EDPII
1 50 100 60 min
1
1
0
100 50
150 min
EDPII
Neo-Cy5

2
1 Neo-Cy5 + Cm
50
0
100 150 min
2 0

Cell counts
102 103 104 105 Neo-Cy5

30 min 2 50 Cy5 Neo-Cy5 + Cm

1
2 0
102 103 104 105
1
30 min 2
Cy5
9 3
2 1 c) 8
EDPI EDPII

n= 240

Cy5 intensity x105


n=
3
1 CFU Log 10 7
2
60 min 3
2
3 d) 6

5
Figure 5 submitted
1
2 n= 149
n= 86
n= 240 4 0
Cy5 intensity x105

Cy5 intensity

n= 217 n= 110 n= 216


3
0 50 100
2 60 min 3
n= 174 Time (min)

Figure 5 submitted
2
Cy5 intensity 104

1 EDPI 0.35

n= 190
1
Membrane 0.65

0 0
0 20 40
Time (min)
Time (min)
Figure 5. EDPI-mediated uptake can be distinguished from EDPII-mediated uptake. a) Representative brightfield and TIRF microscopy
images of cells after 2, 30 and 60 min of exposure to 32 µM Neo-Cy5. Scale bar: 5 µm b) E. coli cells treated with 32 µM Neo-Cy5 with or
without 25 µg/mL of chloramphenicol were incubated for the times indicated. At each time point, cells were filtered, washed, and analyzed
by FACS. Cell counts were normalized to 100%. Weak cytoplasmic uptake, which corresponded to EDPI was detected as an upper shift of
the blue population compared to the red population. The stronger shift corresponding to EDPII levels of uptake is highlighted by an arrow.
c) Killing curve obtained for E. coli with monitoring of Neo-Cy5 uptake. Cells were grown exponentially at 37°C in MOPS medium con-
taining glucose to an OD600 of 0.2, diluted 10 times in fresh medium before addition of Neo-Cy5. At different time points, cells were col-
lected and filtered to remove drug excess. Cells were plated on agar pads for fluorescence microscopy (Figure S14) and fluorescence quan-
tification to generate panel d and serial dilutions were made and plated on LB agar for c.f.u. counting. The kinetic of Neo-Cy5 uptake on
the cell population is represented by the sum of the single-cell analysis of fluorescence signals displayed in panel d. The rapid and linear
EDPII phase is highlighted by a dashed line. d) Single-cell quantification of Neo-Cy5 accumulation as a function of time. Red lines high-
light the medians, boxes show 75/25%, and vertical dashed lines indicate 90/10% of the maxima.
dependent on the neomycin moiety demonstrating that this attention for the development of novel, potent, and less toxic
type of conjugate can be widely used to report on aminogly- drugs to address the pressing societal problem of multidrug-
coside uptake. In addition, these dyes have spectral proper- resistant (MDR) infectious disease.60,61 The approach com-
ties that make them suitable for fluorescence microscopy. bining fluorescence microscopy and FACS should provide a
The Neo-Cy5 conjugate is active against Gram-positive and framework for future antibiotic-cell interaction studies and
negative strains with the exception of Pseudomonas aeru- facilitate the development of novel aminoglycosides.
ginosa in rich medium. This pathogen is known to resist to
multiple drugs owing to a membrane of low permeability,
METHODS
the expression of multidrug efflux pumps and its adaptation
to the presence of antibiotics. The efflux pump MexX- Chemicals. Cy5 and Cy3 NHS-esters were purchased from
MexY-OprM plays a role in intrinsic resistance to aminogly- General Electric Healthcare or Lumiprobe GmbH. 3,5-
cosides.52–54 The pump has moderate drug specificity as Difluoro-4-hydroxybenzylidene imidazolinone was pur-
aminoglycosides, macrolides and tetracycline are sub- chased from Lucerna Technologies. 2,2,2-trifluoro-N-(2-
strates.55 Structural data obtained for this type of pumps sulfanylethyl)acetamide was obtained from Merck or pre-
provided details on their architecture and revealed indeed pared following the reported procedure.62
multiple binding pockets for various drugs.56–58 It remains to Labeling of neomycin with cyanine fluorophores. A de-
be investigated if the presence of the fluorescent dye will rivative of neomycin containing 2-N-
affect the efflux mechanism in Pseudomonas aeruginosa. trifluoroacetamidoethylthio in place of a hydroxyl group in
Using Neo-Cy5 conjugate, we devised a method for accurate ring III was used for functionalization. Other amino groups
monitoring of aminoglycoside uptake. Membrane binding of were protected by hexa-N-tert-butyloxycarbonyl groups for
Neo-Cy5 was found to account for a large fraction of the selective reactivity at the new 5” NH2 position. The deriva-
fluorescence signal. We showed that we could remove the tive was used to conjugate neomycin to NHS-ester dyes.
excess of drug and the weakly bound molecules, which oth- Selective deprotection of the 5” position was performed in
erwise masked the real levels of uptake, by filtration and ammonium hydroxide in methanol, and the unique free ami-
washing. Using TIRF microscopy we ensured the validity of no group was reacted with Cy5 or Cyanine 5 monofunctional
FACS observations. Using FACS, we monitored drug bind- NHS-ester. Reactions were monitored by analytical thin-
ing and intracellular accumulation. Neo-sulfonated-cyanine layer chromatography (TLC) on silica gel 60 F254 plates
derivatives did not suffer from dye permeability drawbacks (0.25 mm). The Cy5 and Cyanine 5 conjugates were purified
observed for dyes such as Texas Red.35 on TLC plates (Silica gel 60, Merck), eluted in ethanol, and
deprotected with trifluoroacetic acid (90%) to remove bu-
Aminoglycosides are bound to the bacterial cell membrane
tyloxycarbonyl groups.
and accumulate in the cytoplasm of bacterial cells through
EDPI and EDPII. The membrane-bound and EDPII compo- Purification of ribosomes. Tightly coupled 70S ribosomes
nents accounted for a large fraction of the total levels of from E. coli MRE600 were prepared as described.63,64 Brief-
Neo-Cy5 tightly associated with E. coli cells. Levels of Neo- ly, cells were grown in exponential phase and when the
Cy5 resulting from EDPI-mediated internalization were low OD600 reached 0.3, cells were collected by centrifugation.
even though it is this fraction that results in the deleterious Cells were lysed by passage through a module/piston ho-
effects of aminoglycosides on cells. The loss of membrane mogenizing system pre-cooled to -80 °C that maintains the
integrity results in the onset of strong diffusive entry of the material in a frozen state. The piston was actuated with a
drug during EDPII.25,27 Carver hydraulic press. Ribosomes were isolated by several
centrifugation steps followed by a sucrose gradient (10-40%)
The data reported here illustrate how fluorescent derivatives
fractionation. The fraction containing 70S particles was
of an aminoglycoside enable a robust characterization of the
collected using a density gradient fraction system (Teledyne
three components of uptake: membrane binding, EDPI, and
Isco), pelleted, and resuspended in 50 mM Tris-HCl (pH
EDPII. EDPII-mediated uptake is a consequence of the mis-
7.6), 10 mM MgCl2, 100 mM NH4Cl, 6 mM 2-
coding activity of the drug that accumulated via EDPI. As
mercaptoethanol. Aliquots were stored at -80 °C after flash
miscoding levels can vary between bacterial strains and
freezing in liquid nitrogen. The ribosome concentration was
between mutants of a single strain, EDPII accumulation may
obtained by measuring the absorption at 260 nm; we as-
only partially reflect EDPI accumulation. Thus, it is critical
sumed that there are 23 pmol ribosomes per A260 unit.
that the three components of aminoglycoside uptake be eval-
uated. We expect that the method described here will be Footprinting of Neo-Cy5 on 70S ribosomes. The concen-
applied to characterize aminoglycoside uptake into bacteria tration of Neo-Cy5 stock solution was estimated from the
to facilitate a better understanding of the mechanisms of absorbance at 649 nm measured in 10 mM Tris-HCl, pH 7.5.
action of these drugs and their next generation derivatives. Chemical probing of Neo-Cy5-ribosome complexes was
Interestingly, the group of Y. Tor recently investigated the performed as described.65 We used the RNA base-specific
uptake of several aminoglycoside-Cy3 conjugates including reagent dimethyl sulfate (DMS) which methylates positions
neomycin-Cy3 and guanidinoneomycin-Cy3 in Chinese N1 of adenines and N3 of cytidines as well as position N7 of
hamster ovarian cells using fluorescence microscopy and guanines.66 Modification reactions (100 µl) with 70S parti-
FACS.59 Guanidinoglycosides-Cy3 conjugates entered cells cles (10 pmol) were performed in a buffer containing 80 mM
better than their parent aminoglycosides. This work together potassium cacodylate pH 7.2, 100 mM ammonium chloride,
with our results emphasize the usefulness of Cyanine- 20 mM magnesium chloride, 1 mM dithiothreitol and 0.5
aminoglycoside conjugates for further studies of their cellu- mM EDTA. In brief, 1 µl of (DMS) (1:10 dilution in 95%
lar uptake by eukaryotic and prokaryotic cells. This class of ethanol) was added to a 50-µL reaction mixture. After incu-
antibiotics, including neomycin, has recently drawn strong bation at 37 °C for 10 minute reactions were stopped by
addition of 25 µL of 6.7% (v/v) β-mercaptoethanol in 280
mM sodium acetate (pH 5.4) and 150 µL of ethanol followed at 37 °C with or without 30 µM carbonyl cyanide m-
by rapid mixing. After precipitation, the pellets were resus- chlorophenyl hydrazone (CCCP). The number of live cells at
pended in 10 µL of H2O. the time of CCCP addition were estimated as 5x107
Sites of modification were analyzed by fluorescent primer c.f.u./mL. After a 4.5-h incubation at 37 °C with neomycin
extension as previously described.67 A Cy5-labeled fluores- or Neo-Cy5, cell survival was measured in c.f.u./mL.
cent DNA primer was purchased from MWG Biotech. Re- Neo-Cy5 uptake. For the liquid culture, MOPS-G was used.
verse transcription reactions were performed in 10 µL of 50 Overnight cultures were diluted to an OD600 of 0.037 with
mM Tris-HCl, pH 8.3, 75 mM KCl, 3 mM MgCl2 10 mM fresh medium and grown until the OD600 reached 0.2. Typi-
DTT with 0.25 mM each dNTP. SuperScript II (Invitrogen, cally, 0.5 µL of Neo-Cy5 dissolved in milliQ water was
0.5 µL 100 units) was added and reactions were incubated at added to 5 µL of the cell suspension (diluted 10 times). Cells
45 °C for 40 min. For sequencing samples, 1.25 mM of were incubated in a heat block at 37 °C without shaking. To
ddATP, ddCTP, ddGTP, or ddTTP was added to individual remove Neo-Cy5 not tightly bound or internalized, the sam-
reverse transcription reactions of each sample. Reactions ples were filtered through 0.45 µm-pore-size HAWP mixed
were stopped by ethanol precipitation. cDNA samples were cellulose ester (MCE) membranes (Merck Millipore) mount-
dissolved in 10 µL loading solution containing 7 M urea. A ed on a micro-sample filtration Minifold II (Schleicher &
small aliquot (1.5 to 3 µL) of each sample was loaded on an Schuell) and washed three times with 100 µL MOPS-G pre-
8% polyacrylamide 7 M urea gel for separation of cDNAs. warmed to 37 °C. Immediately after filtration, the section of
Gels were analyzed on a Typhoon imaging system. Molecu- the membrane containing cells was placed into an Eppendorf
lar graphics were generated using PyMOL(TM) 2.0.1. tube containing MOPS-G, and cells were recovered by gentle
Miscoding assay. Miscoding was assessed in a gain-of- up and down pipetting. Cells were immediately placed on
function assay using firefly and Renilla luciferases. DNA 1% agarose pad for imaging by fluorescence microscopy or
reporter plasmids pT7 hFluc WT, pT7 hFluc H245R(CGC), analysis by FACS.
and pT7 hRluc WT for in vitro translation assay were pro- Fluorescence microscopy. Fluorescence images were taken
vided by Erik Böttger and Dimitri Scherbakov (University of with an EMCCD camera (Photometrics) through a 63X total
Zurich).5 Wild-type and mutant DNAs were used for in vitro internal reflection fluorescence (TIRF) objective (Zeiss, NA
translation reactions together with the Renilla luciferase 1.43) mounted on an inverted Zeiss Axio Observer Z1 mi-
DNA, which served as an internal control. In vitro translation croscope. Image acquisition was done with the Metamorph
reactions were performed using the PURExpress system software package (Molecular Devices). Cy5 illumination was
(New England BioLabs). The premix for one reaction con- performed using a 642-nm laser (0.002 kW/cm2, Roper Sci-
tained 2 µL solution A, 1.5 µL solution B, 5 nM DNA tem- entific), and a filter set was used for fluorescence excitation
plates, and 1 unit/µL of SUPERase·In™ RNase Inhibitor and emission (Chroma Technology; ET 532/640 nm Laser
(Invitrogen). The reaction was begun by addition of 4 µL of Dual Band; excitation filter 530/20 nm and 638/25 nm, di-
the premix lacking aminoglycosides to 1 µL of aminoglyco- chroic mirror 585/70 nm and 650 nm (long pass), and emis-
side dilution in 0.2 ml thin-walled tubes on ice. Synthesis sion filter 580/70 nm and 700/100 nm). Neo-Cy5 uptake was
was performed at 37 °C for 35 min in a thermocycler (lid typically observed with an electron-multiplying gain of 1 or
temperature 98 °C) and stopped by placing samples on ice. 50 and an exposure of 10 to 100 ms. Spinach aptamer bound
Luciferase activity was measured using the Dual-Luciferase to 3,5-difluoro-4-hydroxybenzylidene imidazolinone was
Reporter Assay System (Promega). Luciferase assay reagent excited with a mercury lamp (excitation 470 ± 20 nm, emis-
(50 µl) was added to each sample, and the samples were sion 525 ± 25 nm). Image analyses were performed using
mixed and transferred to a white 96-well plate (Greiner, ImageJ and microbeJ.68
catalog number 655075) for bioluminescence quantification FACS analysis. To measure Cy5 fluorescence, cells were
using an Infinite M200 microplate reader (Tecan). Mis- analyzed with a BD Accuri C6 flow cytometer (BD Biosci-
coding was quantified by calculating the mutant fire- ences). For each experiment, more than 100 000 events were
fly/Renilla luciferase activity compared with the wild-type recorded. Excitation was performed with a 640-nm diode
firefly/Renilla luciferase activity. laser (14.7 mW), and emission was detected through a
Minimum inhibitory concentration measurement. The 675/25 nm band pass filter. Data were analyzed with Kaluza
minimum inhibitory concentrations (MICs) were determined 2.1 (Beckman Coulter) and FlowLogic 7.2. Binding affinity
using a liquid culture assay according to EUCAST guide- experiments were analyzed with the software GraphPad
lines. Aliquots of an overnight culture of E. coli MG1655 Prism 8.2.1. Data were fitted to a simple saturation curve.
were diluted to have an inoculum of 5.5x105 colony-forming
units per mL (c.f.u./mL) into the medium containing 2-fold ASSOCIATED CONTENT
serial dilutions of antibiotics. Growth was measured by
Supporting Information
turbidity at each concentration during 18 h of incubation at
37 °C. Measurements were performed on 200 µL samples in The Supporting Information is available free of charge on the
a 96-well microtiter plate with transparent lid using an Infi- ACS Publications website.
nite M200PRO (TECAN). For Neo-Cy5, turbidity was
Supporting methods, 14 figures and one table showing addition-
measured at 400 nm or 500 nm instead of 600 nm to mini-
al results (PDF).
mize Cy5 excitation.
Treatment of cells with carbonyl cyanide m-chlorophenyl
hydrazone. E. coli MG1655 cells from an overnight culture AUTHOR INFORMATION
in MOPS supplemented with 0.4 % (w/v) glucose (MOPS-G)
were diluted into the same medium. Cells were grown for 2 h Corresponding Author
* E-mail: dominique.fourmy@i2bc.paris-saclay.fr. 2012, 109 (27), 10984–10989.
Present Addresses (6) Sato, R.; Tanigawara, Y.; Kaku, M.; Aikawa, N.; Shimizu,
† Institute for Environmental and Gender-Specific Medicine, K. Pharmacokinetic-Pharmacodynamic Relationship of Arbeka-
Juntendo University Graduate School of Medicine, Chiba 279- cin for Treatment of Patients Infected with Methicillin-Resistant
0021, Japan. Staphylococcus Aureus. Antimicrob. Agents Chemother. 2006,
50 (11), 3763–3769.
Author Contributions
S.Y. and D.F. designed the research strategy. M.S.A., M.O. and (7) Zhanel, G. G.; Lawson, C. D.; Zelenitsky, S.; Findlay, B.;
D.F performed the microscopy. M.S.A. performed the survival Schweizer, F.; Adam, H.; Walkty, A.; Rubinstein, E.; Gin, A. S.;
assays. M.S.A. and D.F performed the FACS and M.R. helped Hoban, D. J.; Lynch, J. P.; Karlowsky, J. A. Comparison of the
with analysis. M.O, S.Y. and D.F. performed the biochemistry. Next-Generation Aminoglycoside Plazomicin to Gentamicin,
M.C. measured MIC in rich medium on Gram positive and Tobramycin and Amikacin. Expert Rev Anti Infect Ther 2012,
negative strains and in minimal medium on pathogenic strains. 10 (4), 459–473.
M.S.A., M.O. and S. Y. performed other MIC experiments. D.F.
performed the chemical synthesis and M.P.H. the NMR and LC- (8) Borovinskaya, M. A.; Pai, R. D.; Zhang, W.; Schuwirth, B.
MS. D.F. wrote the manuscript. All authors have given approval S.; Holton, J. M.; Hirokawa, G.; Kaji, H.; Kaji, A.; Cate, J. H.
to the final version of the manuscript. Structural Basis for Aminoglycoside Inhibition of Bacterial
Ribosome Recycling. Nat Struct Mol Biol 2007, 14 (8), 727–
ACKNOWLEDGMENTS 732.
We wish to thank Marc Mirande, Soufian Ouchane, Anne-Soisig
Steunou, J. Dubois, Stéphanie Bury-Moné for use of equipment. (9) Campuzano, S.; Vazquez, D.; Modolell, J. Functional Inter-
We wish to thank David Buisson for help with the LC-MS. We action of Neomycin B and Related Antibiotics with 30S and 50S
wish to thank Christophe Beloin, Suzana Salcedo, Frédéric Ribosomal Subunits. Biochem Biophys Res Commun 1979, 87
Boccard, Viginia Lioy, Etienne Dervyn, Jean-Luc Pernodet, (3), 960–966.
Philippe Bouloc for strains and Isabelle Broutin for helpful
discussions. The authors dedicate this paper to the memory of (10) Moazed, D.; Noller, H. F. Interaction of Antibiotics with
their colleague Jean-Louis Fourrey. This work was supported by Functional Sites in 16S Ribosomal RNA. Nature 1987, 327
the Centre National de la Recherche Scientifique (CNRS) to (6121), 389–394.
D.F. and S.Y. M.O. was supported by JASSO, a fellowship from
the French embassy in Japan “Bourse du gouvernement fran- (11) Davies, J.; Gorini, L.; Davis, B. D. Misreading of RNA
çais” and a fellowship from FRM (Fondation pour la recherche Codewords Induced by Aminoglycoside Antibiotics. Mol Phar-
Médicale-FDT20140931068). M.S.A. was supported by a PhD macol 1965, 1 (1), 93–106.
fellowship from the French Ministère de l’Enseignement Supé-
rieur et de la Recherche (MESR). (12) Carter, A. P.; Clemons, W. M.; Brodersen, D. E.; Morgan-
Warren, R. J.; Wimberly, B. T.; Ramakrishnan, V. Functional
REFERENCES Insights from the Structure of the 30S Ribosomal Subunit and
Its Interactions with Antibiotics. Nature 2000, 407 (6802), 340–
348.

(1) Jackson, J.; Chen, C.; Buising, K. Aminoglycosides: How (13) Demeshkina, N.; Jenner, L.; Westhof, E.; Yusupov, M.;
Should We Use Them in the 21st Century? Curr. Opin. Infect. Yusupova, G. A New Understanding of the Decoding Principle
Dis. 2013, 26 (6), 516–525. on the Ribosome. Nature 2012, 484 (7393), 256–259.

(2) Takahashi, Y.; Igarashi, M. Destination of Aminoglycoside (14) Fourmy, D.; Recht, M. I.; Blanchard, S. C.; Puglisi, J. D.
Antibiotics in the “Post-Antibiotic Era.” J. Antibiot. 2018, 71, Structure of the A Site of Escherichia Coli 16S Ribosomal RNA
4–14. Complexed with an Aminoglycoside Antibiotic. Science 1996,
274 (5291), 1367–1371.
(3) Aggen, J. B.; Armstrong, E. S.; Goldblum, A. A.; Dozzo, P.;
Linsell, M. S.; Gliedt, M. J.; Hildebrandt, D. J.; Feeney, L. A.; (15) Vicens, Q.; Westhof, E. Crystal Structure of Paromomycin
Kubo, A.; Matias, R. D.; Lopez, S.; Gomez, M.; Wlasichuk, K. Docked into the Eubacterial Ribosomal Decoding A Site. Struc-
B.; Diokno, R.; Miller, G. H.; Moser, H. E. Synthesis and Spec- ture 2001, 9 (8), 647–658.
trum of the Neoglycoside ACHN-490. Antimicrob. Agents
Chemother. 2010, 54 (11), 4636–4642. (16) Yoshizawa, S.; Fourmy, D.; Puglisi, J. D. Structural Origins
of Gentamicin Antibiotic Action. Embo J 1998, 17 (22), 6437–
(4) Huth, M. E.; Han, K.-H.; Sotoudeh, K.; Hsieh, Y.-J.; Effertz, 6448.
T.; Vu, A. A.; Verhoeven, S.; Hsieh, M. H.; Greenhouse, R.;
Cheng, A. G.; Ricci, A. J. Designer Aminoglycosides Prevent (17) Cabanas, M. J.; Vazquez, D.; Modolell, J. Inhibition of
Cochlear Hair Cell Loss and Hearing Loss. J. Clin. Invest. 2015, Ribosomal Translocation by Aminoglycoside Antibiotics. Bio-
125 (2), 583–592. chem Biophys Res Commun 1978, 83 (3), 991–997.

(5) Matt, T.; Ng, C. L.; Lang, K.; Sha, S.-H.; Akbergenov, R.; (18) Misumi, M.; Nishimura, T.; Komai, T.; Tanaka, N. Interac-
Shcherbakov, D.; Meyer, M.; Duscha, S.; Xie, J.; Dubbaka, S. tion of Kanamycin and Related Antibiotics with the Large Sub-
R.; Perez-Fernandez, D.; Vasella, A.; Ramakrishnan, V.; unit of Ribosomes and the Inhibition of Translocation. Biochem
Schacht, J.; Böttger, E. C. Dissociation of Antibacterial Activity Biophys Res Commun 1978, 84 (2), 358–365.
and Aminoglycoside Ototoxicity in the 4-Monosubstituted 2-
Deoxystreptamine Apramycin. Proc. Natl. Acad. Sci. U.S.A. (19) Hirokawa, G.; Kiel, M. C.; Muto, A.; Selmer, M.; Raj, V.
S.; Liljas, A.; Igarashi, K.; Kaji, H.; Kaji, A. Post-Termination cin Kills Antibiotic-Resistant Bacteria. Cell Metab. 2015, 21 (2),
Complex Disassembly by Ribosome Recycling Factor, a Func- 249–262.
tional TRNA Mimic. Embo J 2002, 21 (9), 2272–2281.
(34) Stone, M. R. L.; Butler, M. S.; Phetsang, W.; Cooper, M.
(20) Hirokawa, G.; Kaji, H.; Kaji, A. Inhibition of Antiassocia- A.; Blaskovich, M. A. T. Fluorescent Antibiotics: New Research
tion Activity of Translation Initiation Factor 3 by Paromomycin. Tools to Fight Antibiotic Resistance. Trends Biotechnol. 2018,
Antimicrob Agents Chemother 2007, 51 (1), 175–180. 36 (5), 523–536.

(21) Wallace, B. J.; Tai, P. C.; Herzog, E. L.; Davis, B. D. Par- (35) Allison, K. R.; Brynildsen, M. P.; Collins, J. J. Metabolite-
tial Inhibition of Polysomal Ribosomes of Escherichia Coli by Enabled Eradication of Bacterial Persisters by Aminoglycosides.
Streptomycin. Proc Natl Acad Sci U S A 1973, 70 (4), 1234– Nature 2011, 473 (7346), 216–220.
1237.
(36) Cinquin, B.; Maigre, L.; Pinet, E.; Chevalier, J.; Stavenger,
(22) Foster, C.; Champney, W. S. Characterization of a 30S R. A.; Mills, S.; Réfrégiers, M.; Pagès, J.-M. Microspectromet-
Ribosomal Subunit Assembly Intermediate Found in Escherich- ric Insights on the Uptake of Antibiotics at the Single Bacterial
ia Coli Cells Growing with Neomycin or Paromomycin. Arch Cell Level. Sci Rep 2015, 5, 17968.
Microbiol 2008, 189 (5), 441–449.
(37) Vergalli, J.; Dumont, E.; Pajović, J.; Cinquin, B.; Maigre,
(23) Mehta, R.; Champney, W. S. 30S Ribosomal Subunit As- L.; Masi, M.; Réfrégiers, M.; Pagés, J.-M. Spectrofluorimetric
sembly Is a Target for Inhibition by Aminoglycosides in Esche- Quantification of Antibiotic Drug Concentration in Bacterial
richia Coli. Antimicrob Agents Chemother 2002, 46 (5), 1546– Cells for the Characterization of Translocation across Bacterial
1549. Membranes. Nat Protoc 2018, 13 (6), 1348–1361.

(24) Sykes, M. T.; Shajani, Z.; Sperling, E.; Beck, A. H.; Wil- (38) Dumont, E.; Vergalli, J.; Conraux, L.; Taillier, C.; Vassort,
liamson, J. R. Quantitative Proteomic Analysis of Ribosome A.; Pajovic, J.; Réfrégiers, M.; Mourez, M.; Pagès, J.-M. Anti-
Assembly and Turnover in Vivo. J Mol Biol 2010, 403 (3), 331– biotics and Efflux: Combined Spectrofluorimetry and Mass
345. Spectrometry to Evaluate the Involvement of Concentration and
Efflux Activity in Antibiotic Intracellular Accumulation. J.
(25) Davis, B. D. Mechanism of Bactericidal Action of Amino- Antimicrob. Chemother. 2019, 74 (1), 58–65.
glycosides. Microbiol Rev 1987, 51 (3), 341–350.
(39) Hernandez Linares, A.; Fourmy, D.; Fourrey, J.-L.;
(26) Hancock, R. E. Aminoglycoside Uptake and Mode of Loukaci, A. Introduction of a Substituent at the 5"-Position of
Action-with Special Reference to Streptomycin and Gentamicin. N-Boc Neomycin B under Mitsunobu Reaction Conditions. Org.
II. Effects of Aminoglycosides on Cells. J. Antimicrob. Biomol. Chem. 2005, 3 (11), 2064–2066.
Chemother. 1981, 8 (6), 429–445.
(40) Dempsey, G. T.; Vaughan, J. C.; Chen, K. H.; Bates, M.;
(27) Taber, H. W.; Mueller, J. P.; Miller, P. F.; Arrow, A. S. Zhuang, X. Evaluation of Fluorophores for Optimal Perfor-
Bacterial Uptake of Aminoglycoside Antibiotics. Microbiol Rev mance in Localization-Based Super-Resolution Imaging. Nat
1987, 51 (4), 439–457. Methods 2011, 8 (12), 1027–1036.

(28) Bryan, L. E.; Kwan, S. Mechanisms of Aminoglycoside (41) Recht, M. I.; Douthwaite, S.; Puglisi, J. D. Basis for Pro-
Resistance of Anaerobic Bacteria and Facultative Bacteria karyotic Specificity of Action of Aminoglycoside Antibiotics.
Grown Anaerobically. J. Antimicrob. Chemother. 1981, 8 Suppl Embo J 1999, 18 (11), 3133–3138.
D, 1–8.
(42) Pfister, P.; Hobbie, S.; Brull, C.; Corti, N.; Vasella, A.;
(29) Damper, P. D.; Epstein, W. Role of the Membrane Poten- Westhof, E.; Bottger, E. C. Mutagenesis of 16S RRNA C1409-
tial in Bacterial Resistance to Aminoglycoside Antibiotics. G1491 Base-Pair Differentiates between 6’OH and 6’NH3+
Antimicrob. Agents Chemother. 1981, 20 (6), 803–808. Aminoglycosides. J Mol Biol 2005, 346 (2), 467–475.

(30) Eisenberg, E. S.; Mandel, L. J.; Kaback, H. R.; Miller, M. (43) Perez-Fernandez, D.; Shcherbakov, D.; Matt, T.; Leong, N.
H. Quantitative Association between Electrical Potential across C.; Kudyba, I.; Duscha, S.; Boukari, H.; Patak, R.; Dubbaka, S.
the Cytoplasmic Membrane and Early Gentamicin Uptake and R.; Lang, K.; Meyer, M.; Akbergenov, R.; Freihofer, P.; Vaddi,
Killing in Staphylococcus Aureus. J. Bacteriol. 1984, 157 (3), S.; Thommes, P.; Ramakrishnan, V.; Vasella, A.; Böttger, E. C.
863–867. 4’-O-Substitutions Determine Selectivity of Aminoglycoside
Antibiotics. Nat Commun 2014, 5, 3112.
(31) Davis, B. D.; Chen, L. L.; Tai, P. C. Misread Protein Cre-
ates Membrane Channels: An Essential Step in the Bactericidal (44) Perzynski, S.; Cannon, M.; Cundliffe, E.; Chahwala, S. B.;
Action of Aminoglycosides. Proc Natl Acad Sci U S A 1986, 83 Davies, J. Effects of Apramycin, a Novel Aminoglycoside Anti-
(16), 6164–6168. biotic on Bacterial Protein Synthesis. Eur. J. Biochem. 1979, 99
(3), 623–628.
(32) Hancock, R. E. Aminoglycoside Uptake and Mode of
Action with Special Reference to Streptomycin and Gentamicin. (45) Hancock, R. E.; Farmer, S. W.; Li, Z. S.; Poole, K. Interac-
I. Antagonists and Mutants. J. Antimicrob. Chemother. 1981, 8 tion of Aminoglycosides with the Outer Membranes and Puri-
(4), 249–276. fied Lipopolysaccharide and OmpF Porin of Escherichia Coli.
Antimicrob Agents Chemother 1991, 35 (7), 1309–1314.
(33) Peng, B.; Su, Y.-B.; Li, H.; Han, Y.; Guo, C.; Tian, Y.-M.;
Peng, X.-X. Exogenous Alanine and/or Glucose plus Kanamy- (46) Okuda, M.; Fourmy, D.; Yoshizawa, S. Use of Baby Spin-
ach and Broccoli for Imaging of Structured Cellular RNAs. (58) Murakami, S.; Nakashima, R.; Yamashita, E.; Matsumoto,
Nucleic Acids Res. 2017, 45 (3), 1404–1415. T.; Yamaguchi, A. Crystal Structures of a Multidrug Transporter
Reveal a Functionally Rotating Mechanism. Nature 2006, 443
(47) Jörgensen, F.; Kurland, C. G. Death Rates of Bacterial (7108), 173–179.
Mutants. FEMS Microbiology Letters 1987, 40 (1), 43–46.
(59) Hadidi, K.; Wexselblatt, E.; Esko, J. D.; Tor, Y. Cellular
(48) White, J. R.; White, H. L. Streptomycinoid Antibiotics: Uptake of Modified Aminoglycosides. J. Antibiot. 2018, 71,
Synergism by Puromycin. Science 1964, 146 (3645), 772–774. 142–145.

(49) Klainer, A. S.; Russell, R. R. Effect of the Inhibition of (60) Böttger, E. C.; Crich, D. Aminoglycosides: Time for the
Protein Synthesis on the Escherichia Coli Cell Envelope. Anti- Resurrection of a Neglected Class of Antibacterials? ACS Infect
microb. Agents Chemother. 1974, 6 (2), 216–224. Dis 2020, 6 (2), 168-172.

(50) Ezraty, B.; Vergnes, A.; Banzhaf, M.; Duverger, Y.; Hu- (61) Sati, G. C.; Sarpe, V. A.; Furukawa, T.; Mondal, S.; Man-
guenot, A.; Brochado, A. R.; Su, S. Y.; Espinosa, L.; Loiseau, tovani, M.; Hobbie, S. N.; Vasella, A.; Böttger, E. C.; Crich, D.
L.; Py, B.; Typas, A.; Barras, F. Fe-S Cluster Biosynthesis Modification at the 2’-Position of the 4,5-Series of 2-
Controls Uptake of Aminoglycosides in a ROS-Less Death Deoxystreptamine Aminoglycoside Antibiotics To Resist Ami-
Pathway. Science 2013, 340 (6140), 1583–1587. noglycoside Modifying Enzymes and Increase Ribosomal Tar-
get Selectivity. ACS Infect Dis 2019, 5 (10), 1718–1730.
(51) Bryan, L. E.; Elzen, H. M. V. D. Streptomycin Accumula-
tion in Susceptible and Resistant Strains of Escherichia Coli and (62) Jagt, R. B. C.; Gómez-Biagi, R. F.; Nitz, M. Pattern-Based
Pseudomonas Aeruginosa. Antimicrob. Agents Chemother. Recognition of Heparin Contaminants by an Array of Self-
1976, 9 (6), 928–938. Assembling Fluorescent Receptors. Angew. Chem. Int. Ed. Engl.
2009, 48 (11), 1995–1997.
(52) Aires, J. R.; Köhler, T.; Nikaido, H.; Plésiat, P. Involve-
ment of an Active Efflux System in the Natural Resistance of (63) Mazauric, M. H.; Leroy, J. L.; Visscher, K.; Yoshizawa, S.;
Pseudomonas Aeruginosa to Aminoglycosides. Antimicrob. Fourmy, D. Footprinting Analysis of BWYV Pseudoknot-
Agents Chemother. 1999, 43 (11), 2624–2628. Ribosome Complexes. Rna 2009, 15 (9), 1775–1786.

(53) Sobel, M. L.; McKay, G. A.; Poole, K. Contribution of the (64) Powers, T.; Noller, H. F. A Functional Pseudoknot in 16S
MexXY Multidrug Transporter to Aminoglycoside Resistance in Ribosomal RNA. Embo J 1991, 10 (8), 2203–2214.
Pseudomonas Aeruginosa Clinical Isolates. Antimicrob. Agents
Chemother. 2003, 47 (10), 3202–3207. (65) Yoshizawa, S.; Fourmy, D.; Puglisi, J. D. Recognition of
the Codon-Anticodon Helix by Ribosomal RNA. Science 1999,
(54) Westbrock-Wadman, S.; Sherman, D. R.; Hickey, M. J.; 285 (5434), 1722–1725.
Coulter, S. N.; Zhu, Y. Q.; Warrener, P.; Nguyen, L. Y.;
Shawar, R. M.; Folger, K. R.; Stover, C. K. Characterization of (66) Fourmy, D.; Yoshizawa, S. Protein-RNA Footprinting: An
a Pseudomonas Aeruginosa Efflux Pump Contributing to Ami- Evolving Tool. Wiley Interdiscip Rev RNA 2012, 3 (4), 557–
noglycoside Impermeability. Antimicrob. Agents Chemother. 566.
1999, 43 (12), 2975–2983.
(67) Ying, B. W.; Fourmy, D.; Yoshizawa, S. Substitution of the
(55) Li, X.-Z.; Nikaido, H. Efflux-Mediated Drug Resistance in Use of Radioactivity by Fluorescence for Biochemical Studies
Bacteria. Drugs 2004, 64 (2), 159–204. of RNA. Rna 2007, 13 (11), 2042–2050.

(56) Monlezun, L.; Phan, G.; Benabdelhak, H.; Lascombe, M.- (68) Ducret, A.; Quardokus, E. M.; Brun, Y. V. MicrobeJ, a
B.; Enguéné, V. Y. N.; Picard, M.; Broutin, I. New OprM Struc- Tool for High Throughput Bacterial Cell Detection and Quanti-
ture Highlighting the Nature of the N-Terminal Anchor. Front tative Analysis. Nat Microbiol 2016, 1 (7), 16077.
Microbiol 2015, 6, 667.

(57) Nakashima, R.; Sakurai, K.; Yamasaki, S.; Nishino, K.;


Yamaguchi, A. Structures of the Multidrug Exporter AcrB
Reveal a Proximal Multisite Drug-Binding Pocket. Nature 2011,
480 (7378), 565–569.
For Table of Contents Only
Fluorescent aminoglycoside antibiotics and methods for accurately monitoring uptake by bacteria

Mahnaz Sabeti Azad, Maho Okuda, Mélina Cyrenne, Mickael Bourge, Marie-Pierre Heck, Satoko Yo-
shizawa, and Dominique Fourmy.

brief synopsis, describing the graphic and explaining how the manuscript relates to sustainability:

Fluorescently labeled aminoglycoside derivatives with uptake and bactericidal activities similar to those
of the parent drug were generated for facilitating studies involving aminoglycoside antibiotics. Robust
characterization
Structures corrigées :of the drug uptake is enabled by the method which combines fluorescence microscopy
with fluorescence-activated
Structures corrigées :
cell sorting (FACS) using neomycin coupled to non-permeable cyanine
dyes.
Neomycin
Neomycin

FACS Background
Cell OD600 0.3
Cell OD600 0.03
Neo-Cy5Neo-Cy5 Cell OD600 0.003

Count
Intermediate
Intermediate for
Cy5
NHS Cy5
for coupling NHS Cy5
coupling P % C %
0,3 93,39 6,61
0,03 55,12 44,88
0,003 32,15 66,22
Figure S3. Scheme for generation of Neo-Cy5. The primary amine group that reacts
Neo-Cy5
with the NHS-ester-Cy5 is highlighted by the red circle. Other amino groups are
Aminoglycoside antibiotic Figure 8
Figure S3. protected by hexa-N-tert-butyloxycarbonyl groups.
Scheme for generation of Neo-Cy5. The primary amine group that reacts Neo-Cy5 Neo-Cy3 Okuda et al., 2018
with the NHS-ester-Cy5 is highlighted by the red circle. Other amino groups are
protected by hexa-N-tert-butyloxycarbonyl groups.
Supporting Information

Fluorescent aminoglycoside antibiotics and methods for accurately monitoring
uptake by bacteria

Mahnaz Sabeti Azad†, Maho Okuda†,‡, Mélina Cyrenne†, Mickael Bourge†, Marie-Pierre
Heck§, Satoko Yoshizawa†, and Dominique Fourmy*,†.
† Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC),

91198, Gif-sur-Yvette, France.


§ Université Paris-Saclay, CEA, Service de Chimie Bio-organique et de Marquage, 91191

Gif-sur-Yvette, France.
Corresponding author
*E-mail : dominique.fourmy@i2bc.paris-saclay.fr


17 pages
Supporting methods
14 figures
1 tables:

S1

Supporting methods
Products were verified by 1H and 13C Nuclear Magnetic Resonance spectroscopy (NMR)
or Liquid Chromatography Mass Spectrometry (LC-MS). NMR could not be performed for
Neo-Cy5 as the quantities synthetized were too low.

Synthetic Procedures and characterization data


1,3,2',6',2''',6''' Hexa-N-tert-butyloxycarbonylneomycin 2: To a solution of
commercially available Neomycin sulfate (1 g, 1,4 mmol) in DMF/H2O (20 mL/20 mL)
were added triethylamine (9 mL) and di-tert-butyl dicarbonate (2,1 g, 9,62 mmol, 6.9
eq.). The mixture was heated at 60° C for 5 h. After concentration under vacuum, the
crude was dissolved in EtOAc and washed with brine. The organic layer was recovered
and dried on MgSO4. After concentration and purification of the crude by silicagel
column chromatography (CH2Cl2/MeOH (9/1)) 2 was isolated (876 mg, 52 % yield; oil).
1H NMR (300 MHz, CD OD) d = 5.45 (br, 1H, H1'), 5.25 (br, 1H, H1''), 5.10 (br, 1H, H1'''),
3
4.9-4.8 (m, 2H), 4.2-4.11 (br, 2H), 3.9 (m, 1H), 3.85-3.78 (m, 3H), 3.76-3.55 (m, 5H), 3.52-
3.39 (m, 6H), 3.32-3.22 (m, 5H), 1.90 (br, 1H, Heq), 1.49-1.35 (m, 54H), 1.3 (br, 1H,
Hax);13C NMR (75 MHz, CD3OD) d = 158.8, 158.3, 158.0, 157.8; 110.5, 100.2, 99.2, 87.6,
83.3, 80.6, 78.2, 75.6, 74.3, 72.9, 72.4, 71.5, 69.0, 63.4, 57.1, 53.4, 52.2, 51.7, 42.6, 41.7,
35.8, 28.9. MS (ESI+): m/z calculated for [C53H94N6O25+Na]+: 1237,6, found : 1237,4;
elemental Analysis calculated C, 52.38; H, 7.80; N, 6.92; O, 32.91; found C, 49.95; H, 7.91;
N, 6.55; O, 30.86.



5"-O-triisopropylbenzensulfonyl-1,3,2',6',2"',6"'-hexa-N-tert-butyloxycarbonyl
Neomycin 3: To a solution of compound 2 (500 mg, 0.41 mmol) in pyridine (3 mL) was
added a solution of 2,4,6 triisopropylbenzenesulfonyl chloride (4 g) in pyridine (7 mL).
The solution was stirred at RT for 48 h and was concentrated under vaccuum. The crude
was dissolved in EtOAc and washed with brine. The organic layer was recovered, dried
on MgSO4. After concentration and purification by silicagel column chromatography
(CH2Cl2/MeOH (10/1)) 3 was isolated (432 mg, 71 % yield); 1H NMR (300 MHz, CD3OD)
d = 7.01 (s, 2H, Har), 6.1 (br, 1H, H1'), 5.18 (br, 1H, Heq), 4.9 (br, 1H, H1"'), 4.65-3.79 (m,

S2

8H), 4.3-4.0(m, 2H), 3.8-3.6 (m, 8H), 3.4-3.2 (m, 4H), 2.7 (br, 1H, Heq), 2.92-2.85 (m, 3H),
1.69 (br, 1H, Hax), 1.9-1.3 (m, 55H) . 1.18 (s, 18H); 13C NMR (75 MHz, CD3OD) d = 169.5,
159.4, 158.7, 158.5, 158.2, 157.7, 132.8, 132.4, 130.9, 130.2, 125.5, 111.1, 100.0, 98.6,
87.0, 81.1, 80.8, 80.7, 80.3, 79.2, 75.8, 74.8, 73.2, 72.8, 71.8, 71.2, 69.8, 69.4, 68.8, 68.5,
56.7, 53.7, 52.6, 42.7, 41.6, 35.7, 29.3, 28.9, 25.7, 24.2, 25.0; MS (ESI+): m/z calculated for
[C68H116N6O27S1+Na+H]+ : 1504,7; found : 1504.8, [C68H116N6O27S1+2Na]2+ : 763.35,
found : 763.5.



5"-(2-N-trifluoroacetamidoethylthiol)-1,3,2',6',2"',6"'-Hexa-N-tert-
butyloxycarbonyl-5"'-deoxyneomycin 4: To a solution of sodium ethoxide (4 mL, 3 M
in EtOH) under Argon was added commercially available 2,2,2-trifluoro-N-(2-
mercaptoethyl)acetamide (90 mg, 0.51 mmol) in EtOH (3 mL) and the mixture was
stirred at RT for 15 min. Then was added a solution of compound 3 (220 mg, 0.15 mmol)
in EtOH (3 mL) and the mixture was stirred at RT for 5 h. 20 mL of CH2Cl2 were added,
and then an aqueous solution of sodium phosphate (pH = 6). The organic layer was
separated, dried on MgSO4 and concentrated. The crude was purified by silicagel column
chromatography (CH2Cl2/MeOH (10/1)) to afford 4 (184 mg, 91 % yield); 1H NMR (300
MHz, CD3OD) d = 6.75 (m, 1H), 6.52 (m, 2H), 6.15 (br, 1H, H1'), 5.45 (br, 1H, H1"), 5.21
(br, 1H, H1"'), 4.8-4.3 (m, 6H), 4.28-4.15 (m, 2H), 4.08-4.0 (m, 1H), 3.8-3.75 (m, 2H),
3.72-3.63 (m, 2H), 3.59-3.45 (m, 6H), 3.39-3.22 (m, 4H), 2.81-2.79(m, 2H), 2.76-2.70 (m,
2H), 2.03-1.96 (m, 1H, Heq), 1.78-1.74(m, 1H), 1.52-1.48 (m, 54H+Hax); MS (ESI-TOF):
m/z calculated for [C57H98F3N7O25S1+1H]+ :1371,5 found : 1371,5
[[C57H98F3N7O25S1+1H] : 686.3, found : 686.4.
2+



5"-(2-aminoethylthio-)-1,3,2',6',2"',6"'-Hexa-N-tert-butyloxycarbonyl-5"-
deoxyneomycin 5:
Compound 4 (84 mg, 0.061 mmol) was dissolved in NH4OH solution in MeOH (3 mL,
70%). The solution was strirred at TA for 1 h and then concentrated under vaccum to

S3

afford 5 (36 mg, 46% yield). 1H NMR (300 MHz, CD3OD) d =5.44 (br, 1H, H1'), 5.2 (br,
1H, H1"'), 4.92 (br, 1H, H1"'), 1H), 4.34 (m, 1H), 4.22(m, 1H), 4.05 (m, 1H), 3.9 (m, 2H),
3.75(m, 2H), 3.64-3.12 (m, 15H), 3.0-2.78 (m, 4H), 1.95 (br, 1H, Heq), 1.68 (m, 1H, Hax),
1.64-1.38 (m, 54H); MS (ESI-TOF): m/z calculated for [C55H99N7O24S1+2H]+ :1276,5
found : 1276,06, [[C55H99N7O24S1+2H:]2+ : 638.5 found : 638.45.

Neo-Cy5


The compound 5 was resuspended in 100 µL of dimethyl formamide (DMF). N,N-
Diisopropylethylamine (100 µL) was added to two micromoles of Cy5 (GE Healthcare)
or Cyanine 5 (Lumiprobe) monofunctional NHS-ester previously dissolved in 500 µL
DMF. The sample was next placed for two hours at room temperature with the Boc-
neomycin derivative for coupling. The reaction was monitored by analytical thin-layer
chromatography (TLC) on silica gel 60 F254 plates with eluant:
chloroform/Ethanol/NH4OH 5.6/3.8/0.45 (Figure S6c). Then the sample was dried
under vacuum and kept at – 80°C if necessary. The product was dissolved in methanol
for purification on TLC plates (Silica gel 60, Merck). The silica gel was recovered and
placed in Eppendorf tubes and the product eluted in ethanol. The resin and the solvent
were separated by centrifugation and the elution step repeated 3 times. The sample was
dried and finally, removal of the butyloxycarbonyl groups was performed in
trifluoroacetic acid (90%) for 10 minutes at room temperature.

S4

R21 =

11a, 11b OH
O
H2N
R3
O HN
12a, 12b (R3= H)
HO
HO H2N O N
Paromomycin
H2N
O NH2
O 13a, 13b (R3= CH3 ) O
R1 O
0.01 +M

OH
0.1 +M
0.5 +M

2.5 +M
- DMS

10 +M

Me
0 +M

1 +M

5 +M

HN
UGCA
R2 OH 14a, 14b
O N
NH

a : R 2 = OH G1494 O
O
H2N O
NH2 Me
b : R2 =
HN
HO 15a, 15b
HO O
Manip du 9/03/07 H O N
H
N
Neomycin Neomycin U Paromomycin after Chloro S
extraction O
0.01 +M

0.1 +M
0.5 +M
0.1 +M
0.5 +M

2.5 +M
0.1 +M
0.5 +M

2.5 +M

16b NH2
10 +M
20 +M
50 +M
- DMS


10 +M

1 +M

5 +M
0 +M

1 +M

5 +M

Figure S1. Derivatives of neomycin. UGCA


Sup Figure 7
Table S1 : MIC values for neomycin derivatives against E. coli MG1655 cells grown
in LB medium.
product N° R1 MIC μg/ml
11b OH 5
G1494
12b uracil 10
13b thymine >40
14b thyminylacetyl 20
15b thyminylacetamidoethylthio 5
16b NH2 >40

Gel du 20/03/07

Neomycin Neo-U
0.01 +M

0.5 +M

2.5 +M
0.5 +M

2.5 +M

10 +M
- DMS

10 +M
0 +M

1 +M

5 +M
0 +M

1 +M

5 +M

A1492
G1494

S5


Figure S2. DMS probing of 30S ribosomal subunits bound to neomycin and Neo-U. The
lane marked “- DMS” is a control reaction with no DMS added. Bands corresponding to
Structures corrigées :
nucleotides A1492 and G1494 are indicated.

Neomycin

Neo-Cy5

Intermediate
for NHS Cy5
coupling



Figure S3. Scheme for generation of Neo-Cy5. The primary amine group that reacts
Figure S3. Scheme for generation of Neo-Cy5. The primary amine group that reacts with
with the NHS-ester-Cy5 is highlighted by the red circle. Other amino groups are
the NHS-ester-Cy5 is highlighted by the red circle. Other amino groups are protected by
protected by hexa-N-tert-butyloxycarbonyl groups.
N-tert-butyloxycarbonyl groups.

52 % yield 71 % yield

Neomycin 1

46 % yield
91 % yield

Conditions and reagents :


a) TEA, Boc2O, DMF, 60°C; NH4OH, MeOH; b) TIPBSCl, Py, RT 48 h; c) EtONa, trifluoro-N-(2-mercaptoethyl)acetamide, RT, 5 h; d) NH4OH, MeOH


Figure S4. Synthesis of Boc-Neo-Cy5 from Boc-Neomycin. All steps were performed at
LE 1e rendement
room temperature.
est a 52 % après
calcul

S6

a) b)
rrigées :

6 Neo-Cy5

Conditions and reagents : a) NH4OH, MeOH, b) i) NHS-Cy5, DIEA, DMF; ii) TFA/H2O 9/1

Neo-Cy5Neo-Cy5

I noted a mistake on feb 10, the midle structure is haveving Cy5 GE and final one Cy5 lumiprobe. I put a white box to correct
NHS Cy5
NHS Cy5

Neo-Cy5 (GE Healthcare) Neo-Cy5 (Lumiprobe)



eneration of Neo-Cy5. TheS5.
Figure primary amine group
Synthesis that reacts
of Boc-Neo-Cy5 from Boc-Neomycin. All steps were performed at
is highlighted by the red circle. Other amino groups are
butyloxycarbonyl
room temperature. Below are indicated the two structures of Neo-Cy5 according to the
n of Neo-Cy5.groups.
The primary amine group that reacts
source of Cy5-NHS (GE Healthcare or Lumiprobe). The difference is highlighted by an
lighted by the red circle. Other amino groups are
arrow.

ycarbonyl groups.

S7

Néomycine DF 63

a)

Chemical Formula: C57H98F3N7O25S

MS(ESI-TOF) m/z calcd for [C57H98F3N7O25S +1H]+ = 1371,5 found = 1371,5

Sup Figur
et j'ai aussi son 1H RMN

S8

b) MS obtained using an ESI-Quadripole autopurify (pump : 2545, mass: ZQ2000) mass spectrometer.

MS(ESI-TOF) m/z calcd for [C55H99N7O24S1+2H]+ = 1276,5, found = 1276,06

MH450b2 411 (3.011) Cm (405:432) 1: Scan ES+


100
1276.06 1.91e7
638.45 19132344
17661404
%

588.37
8173135

1275.30
560.31 4674288
4008259

124.07 532.29 667.93


1345984 1400102 1443313
214.18
869.00 914.77 988.67 1273.35 1278.52 1914.23 1953.63
328363 503.92;324765 668.94;85413 1163.41 18765 276617 1394.96 1559.25 1701.16 1839.64 477468 4927
89978 17156 5095 2735 6552 6118 62490 4822
0 m/z
200 300 400 500 600 700 800 900 1000 1100 1200 1300 1400 1500 1600 1700 1800 1900
MH450b2 (0.003) Is (1.00,1.00) C55H99N7O24S 1: Scan ES+
100
116.79 319.42 637.83 1274.65 4.78e12
4778909237248 4778909237248 4778909237248 4778909237248

425.89 638.33 1275.66


3100853665792 3100853665792 3100853665792
%

426.22 638.83 1276.66


1439767068672 1439767068672 1439767068672

426.56 639.33 1277.66


497368662016 497368662016 497368662016

426.89;140700057600 639.83;140700057600 1278.66;140700057600

0 m/z
200 300 400 500 600 700 800 900 1000 1100 1200 1300 1400 1500 1600 1700 1800 1900
MH450b2 411 (3.011) Cm (409:420) 1: Scan ES+
100
1276.09 2.82e7
28204382

638.43
20496154
%

588.41
9886631 1275.32
7899083

560.29
4904662

532.33
1724265 667.80
119.99 214.25 1018006 1032.99 1273.29 1278.50 1447.42 1738.90 1914.28 1958.06
850.85 914.77 981.39 1700.18
407787 200057 460.14;108542 878 1152.63 25037 351592 1394.96 12527 1561.12 4804 900470 5128
73349 19188 4078 566 14634 5949 74418
0 m/z


200 300 400 500 600 700 800 900 1000 1100 1200 1300 1400 1500 1600 1700 1800 1900

S9

c) Loading Neo-Cy5 Neo-Cy3

Neo-Cy5
Cy5-NHS

Neo-Cy3

Cy3-NHS

d) Neo-Cy5

Neo-Cy5
Produit final Neo Cy 5


e)
Sup Figure xx

Neo-Cy5

MS obtained using an ESI-Quadripole autopurify (pump : 2545, mass: ZQ2000) mass spectrometer.

MS(ESI-TOF) m/z calcd for [C57H87N9O19S3-1H]- = 1297,54, found = 1297,56; [C57H87N9O19S3-2H]2-=


648.75, found = 648,37; [C57H87N9O19S3+3H]3+ = 433,67, found = 434,01.


NEO-CY5-S4 513 (3.759) Cm (477:640) 1: Scan ES+


100
434.01 8.67e6

S10

%

Sup
MS(ESI-TOF) m/z calcd for [C57H87N9O19
648.75, found = 648,37; [C57H87N9O19S3

NEO-CY5-S4 513 (3.759) Cm (477:640)
434.01
100
%

214.27

124.18

336.14
650.29

230.79

346.32
438.72 505.10
642.49 651.19
231.61 325.85
130.62
158.26 389.58 460.55 505.92 652.01
251.28 325.13 603.72 691.69 724.86 810.78 866.71
463.52 576.21 777.17 906
0
150 200 250 300 350 400 450 500 550 600 650 700 750 800 850 900

S11


NEO-CY5-S4 497 (3.645) Cm (473:613)
320.04
100
%

640.55

342.84

648.37

137.04
670.80
141.04 671.69

969.78 1297.56
343.70 859.83
159.05 1298.51
968.87
368.18 392.47 435.27 860.79 961.79 1296.77
319.09 682.07 854.87 971.13 1012.55 1073.30
190.03 395.44 504.51 517.28 556.22 639.31 708.22 879.21 1088.461124.48 1282.89 1320.49 1356.92
234.57 453.52 738.47 795.40 912.09 142


150 200 250 300 350 400 450 500 550 600 650 700 750 800 850 900 950 1000 1050 1100 1150 1200 1250 1300 1350 1400



NEO-CY5-S4

2.75e+1
3.68

2.5e+1

2.25e+1

2.0e+1


1.75e+1
1.14
1.5e+1
AU

Figure S6. Characterization of Boc-neomycin derivative and Neo-Cy5. a) LC-MS of 2-N-


1.25e+1 3.94

1.0e+1

trifluoroacetamidoethylthio-neomycin derivative. b) LC-MS of deprotected 2-N-


7.5

5.0

trifluoroacetamidoethylthio-neomycin
2.5

0.0
derivative. c) Analytical thin-layer
0.94

chromatography of the Neo-Cy5 conjugate. After selective deprotection of the 5” position


NEO-CY5-S4
-0.00 0.20 0.40 0.60 0.80 1.00 1.20 1.40 1.60 1.80 2.00 2.20 2.40 2.60 2.80 3.00 3.20 3.40 3.60 3.80 4.00 4.20 4.40 4.60 4.80 5.00 5.20 5.40 5.

of Boc-neomycin, the free amino group was reacted with Cy5 monofunctional NHS-ester 1.09
1.20
1.11 1.17
3.63 3.65

3.67

and the reaction monitored by TLC (silica gel 60 F254 plates; 1.24

chloroform/Ethanol/NH4OH 28%/5.6:3.8:0.45). The results showed that reactions were


3.68

1.28 3.71
5.41 5.6

complete. d) After purification on TLC plates (Silica gel 60, Merck), Neo-Cy5 was eluted
3.89 5.50
3.73
3.80 5.19 5.27 5.32
3.92
%

1.33

and the purity verified again by TLC. c) LC-MS characterization of Neo-Cy5. e) LC-MS of 1.37
1.41 3.02

the Neo-Cy5 conjugate (Cy5 from Lumiprobe, see Figure S5).


0.89 1.42
0.87 2.772.81 2.89 2.99 3.08 3.14 3.273.31 3.51
0.92 1.61
0.12 0.18 1.51 1.72 3.40 3.45
1.65 1.93
0.45 0.62 0.67 0.77
0.30 0.32


0.98

5
-0.00 0.20 0.40 0.60 0.80 1.00 1.20 1.40 1.60 1.80 2.00 2.20 2.40 2.60 2.80 3.00 3.20 3.40 3.60 3.80 4.00 4.20 4.40 4.60 4.80 5.00 5.20 5.40 5.
NEO-CY5-S4
3.76
3.73
3.72 3.79
0.00
3.81

3.59 3.67 3.90

3.92
4.00

S12
5

3.56

3.53

1.30
3.46
0.07 0.11 3.25 3.26 3.283.35
0.19 0.27 0.89 2.96 3.07
0.66 1.06 1.19 1.24 1.65 2.21 2.85 2.86
0.73
90

80
Cy5 Ex
70
Neo-Cy5 Ex
Fluorescence (a.u.)

60

50 Cy5 Em
40
Neo-Cy5 Em
30

20

10

0
400
413
426
439
452
465
478
491
504
517
530
543
556
569
582
595
608
621
634
647
660
673
686
699
712
725
738
751
764
777
790
Wavelength (nm)

Figure S7. Excitation and emission spectra of Cy5 and Neo-Cy5. Data were recorded for
1 μM solutions of Cy5 or Neo-Cy5 in Tris-HCl (pH 7.6). Optical properties are identical.

Neomycin
4
3
Log 10 (survival percentage)

2
1
0
-1
-2
-3
-4
no antibiotic 0.75 0.85 1 1.5 Antibiotic µM
untreated 3,20 -1,06 -1,53 -2,14 -2,83
CCCP (30 µM) 3,10 2,21 1,97 2,08 1,23

Neo-Cy5
4
Log 10 (survival percentage)

3
2
1
0
-1
-2
-3
-4
-5
no antibiotic 32 48 Antibiotic µM
untreated 3,27 1,213199641 -3,863333333
CCCP (30 µM) 3,116 2,949488188 1,116666667

Figure S8. Neomycin and Neo-Cy5-induced cell killing is PMF dependent.
Representative experiments for survival of E. coli MG1655 cells after a 4.5-h treatment
with various concentrations of neomycin and Neo-Cy5 with and without a 2-h pre-
incubation with 30 μM CCCP. Plotted are values relative to untreated control culture.

S13

10000

Fluorescence Neo-Cy5 (a. u.)


9000
8000
7000
6000
5000
4000
3000
2000
1000
0
0 1 2 3 4

Number wash
Figure S9. Excess Neo-Cy5 is removed by filtration. E. coli MG1655 cells (OD600 0.02)
were incubated 5 minutes with 32 µM Neo-Cy5 at 37 °C then filtered through a 0.45-µm
pore-size HAWP membrane. Cells were washed or not with 100 µL MOPS-G medium
sup Figure 1
(pre-warmed to 37 °C), recovered from the filter, and immediately analyzed by FACS.
Okuda et al., 2019
The average of Neo-Cy5 signals for each sample was measured. Three washing steps
were required to remove the Neo-Cy5.


Figure S10. Cy5 does not enter live E. coli cells. Left: Transmitted light image of a field of
E. coli MG1655 cells exposed to 32 µM Cy5 for 1 h at 37 °C. Cells were washed on a 0.45
µm-pore-size HAWP membrane filter. Right: Fluorescence image of the same field of
view obtained with an electron-multiplying gain of 50 and an exposure of 50 ms.

S14


Peripheral Neo-Cy5 and Spinach-ribosome
BF Cy5 Spinach Cy5 + Spinach

Figure S11. Neo-Cy5 does not localize with ribosomes after a 5-minute incubation. E.
coli cells expressing Spinach-tagged ribosomes were exposed to Neo-Cy5 and imaged.
Cells were illuminated with a 642-nm laser for Neo-Cy5 excitation and emission was
Sup fig 7
detected at 670 nm. Spinach aptamer bound to DFHBI was excited with a mercury lamp,
revised
and images were acquired with a GFP filter cube (ex. 470 ± 20 nm, em. 525 ± 25 nm).
Brightfield, Neo-Cy5 (red), Spinach (green), and merged images.


Fluorescence intensity (a. u.)

104
Fluorescence intensity (a. u.)

103 104

103

102 102 32 34 36 38
Time (s)

0 20 40 60 80 100 120
Time (s)
Figure S12. Fast kinetics of Neo-Cy5 interaction with the membranes revealed by real–
time FACS experiments. Cells in exponential growth phase (0.3 OD600) were diluted 20-
fold to a volume of 150 µL and placed into the flow cytometer for analysis at 37 °C. FACS
analysis was initiated and, after a delay of 20 s, 500 µL of Neo-Cy5 solution was added
for a final concentration of 0.4 µM. Addition of the drug diluted the cells to 0.003 OD600.
Fluorescence intensities of individual cells are plotted vs. time. Inset represents the
integration of the fluorescence signals within the boxed area. The signals were average
over 200-ms intervals during this 6-s time period.

S15




Figure S13: Neo-Cy5 induces morphological defects in E. coli cells grown overnight in
M9 glucose (0.4 % (w/v)). Bright-field images are displayed on the right and
fluorescence images on the left. At a concentration of 4 µM, cells are longer and at 8 µM,
foci are observed at poles.

S16

BF TIRF TIRF (Cy5)
(Cy5) adjusted contrast

2 min

5 min

10 min

20 min

40 min

60 min

80 min

120 min Sup F





Figure S14: Time-course experiment of changes in Neo-Cy5 accumulation levels. Fast
growing cells (OD600: 0.02) were incubated at 37 °C with Neo-Cy5 (32 µM). At each time
points cells were washed on filter and then observe directly on MOPS-G agarose pad
(Scale bar: 5 µm). Images were scaled to the minimum and maximum pixel values of the
first image t=2mn. For the first 4 time points, contrast was adjusted for better
visualization and displayed on the right column. For each time point, images were
analyzed with microbeJ for quantification of Cy5 signals to generate figure 5 panel d.

S17

You might also like